Next Article in Journal
Ultrathin Graphene Oxide-Based Nanocomposite Membranes for Water Purification
Next Article in Special Issue
Mechanosensitive Ion Channels and Their Role in Cancer Cells
Previous Article in Journal
NucEnvDB: A Database of Nuclear Envelope Proteins and Their Interactions
Previous Article in Special Issue
Regulation of Papillary Muscle Contractility by NAD and Ammonia Interplay: Contribution of Ion Channels and Exchangers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Infarct-Reducing Effect of the δ2 Opioid Receptor Agonist Deltorphin II: The Molecular Mechanism

1
Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634012 Tomsk, Russia
2
Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Chidambaram 608002, India
3
Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India
4
Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi’an 710032, China
5
The Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Pushchino, Russia
*
Author to whom correspondence should be addressed.
Membranes 2023, 13(1), 63; https://doi.org/10.3390/membranes13010063
Submission received: 16 November 2022 / Revised: 28 December 2022 / Accepted: 31 December 2022 / Published: 4 January 2023
(This article belongs to the Special Issue Membrane Permeability and Channels)

Abstract

:
The search for novel drugs for the treatment of acute myocardial infarction and reperfusion injury of the heart is an urgent aim of modern pharmacology. Opioid peptides could be such potential drugs in this area. However, the molecular mechanism of the infarct-limiting effect of opioids in reperfusion remains unexplored. The objective of this research was to study the signaling mechanisms of the cardioprotective effect of deltorphin II in reperfusion. Rats were subjected to coronary artery occlusion (45 min) and reperfusion (2 h). The ratio of infarct size/area at risk was determined. This study indicated that the cardioprotective effect of deltorphin II in reperfusion is mediated via the activation of peripheral δ2 opioid receptor (OR), which is most likely localized in cardiomyocytes. We studied the role of guanylyl cyclase, protein kinase Cδ (PKCδ), phosphatidylinositol-3-kinase (PI3-kinase), extracellular signal-regulated kinase-1/2 (ERK1/2-kinase), ATP-sensitive K+-channels (KATP channels), mitochondrial permeability transition pore (MPTP), NO synthase (NOS), protein kinase A (PKA), Janus 2 kinase, AMP-activated protein kinase (AMPK), the large conductance calcium-activated potassium channel (BKCa-channel), reactive oxygen species (ROS) in the cardioprotective effect of deltorphin II. The infarct-reducing effect of deltorphin II appeared to be mediated via the activation of PKCδ, PI3-kinase, ERK1/2-kinase, sarcolemmal KATP channel opening, and MPTP closing.

1. Introduction

The incidence of acute myocardial infarction and mortality did not decrease in the last decade, despite the increasing use of modern interventional and conservative methods of treatment [1]. Percutaneous coronary intervention (PCI) provides >95% restoration of blood flow in the infarct-related coronary artery, but some patients still die [2]. The use of thrombolysis and PCI in clinical practice has made reperfusion injury of the heart come to the fore, which causes the death of about 50% of cardiomyocytes in the ischemia/reperfusion (I/R) zone [3]. Currently, there are no drugs in clinical practice capable of effectively preventing the reperfusion damage to the heart. Therefore, the search for novel drugs capable of effectively preventing cardiac reperfusion injury is considered a high priority of modern pharmacology and experimental cardiology.
The results of our preliminary studies indicated that OR agonists can prevent not only ischemic, but also reperfusion injury of the heart [4]. There are potential reasons to believe that OR agonists can serve as a prototype for creating a new class of drugs that increase cardiac resistance to reperfusion. In 2009, we found that the selective δ2 OR agonist deltorphin II could prevent cardiac I/R injury when administered before ischemia [5]. In 2021, we found that deltorphin II can prevent reperfusion injury of the heart [4]. Our studies indicated that the infarct-reducing effect of deltorphin II is mediated via the activation of δ2 OR in cardiomyocytes [4]. However, the molecular mechanism of the cardioprotective effect of deltorphin II in reperfusion remains unknown.
The objective of this research was to study the intracellular signaling mechanism(s) of the cardioprotective effect of deltorphin II in reperfusion.

2. Materials and Methods

2.1. Materials

Male Wistar rats were purchased from the Institute of Cytology and Genetics SB RAS (Novosibirsk) and used in the present study. The animals were housed in 12:12 h light/dark conditions at the temperature of 24 ± 2 °C. The rats received commercial laboratory feed pellets, along with drinking water ad libitum. All procedures were governed by the Directive 2010/63/EU of the European Parliament and the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication No. 85-23, revised 1996). The Ethical Committee of the Cardiology Research Institute of the Tomsk National Research Medical Center granted the approval for this study (Protocol N° 207 dated 23 December 2020).

2.2. Coronary Artery Occlusion and Reperfusion

The rats were anesthetized with chloralose (50 mg/kg, intraperitoneally, Sigma-Aldrich, St. Louis, MO, USA) and connected to a SAR-830 Small Animal Ventilator artificial respirator (CWE, Inc., Ardmore, PA, USA). They underwent thoracotomy, and the pericardium was removed. The ligature of the left descending coronary artery was superimposed 1–2 mm below the left atrial appendage according to a previously published method [6]. Coronary occlusion was verified by ST segment elevation. The right carotid artery was cannulated for measuring blood pressure, which was registered using an SS13L pressure transducer (Biopac System Inc., Goleta, CA, USA) coupled with an MP35 electrophysiological device (Biopac System Inc., Goleta, CA, USA). The same apparatus was used to record the ECG. Quantitative data processing was performed using INSTBSL-W software of Biopac System Inc., (Goleta, CA, USA).
After 45 min of ischemia, the silk ligature was loosened, and the restoration of the blood flow was confirmed by the appearance of epicardial hyperemia. The duration of reperfusion was 2 h.

2.3. Detection of Infarct Size and the Area at Risk

The identification of the necrosis zone and the risk zone was carried out according to the method proposed by Neckar et al. [7]. After reperfusion, the hearts were removed and rinsed with a syringe through the cannulated aorta with a saline solution containing 125 IU/mL of heparin. To determine the area at risk, the ligature was re-tightened, and the myocardium was stained through the aorta with 5% potassium permanganate; after washing, 1 mm thick heart sections perpendicular to the longitudinal axis were prepared. The necrosis zone was isolated from the risk zone by staining with a 1% solution of 2,3,5-triphenyl tetrazolium chloride for 30 min at 37 °C. After staining, the slices were placed in a 10% formaldehyde solution for 1 day. The day after staining, the right ventricle was removed, and left ventricular sections were scanned on both sides with an HP Scanjet G2710 scanner. The area at risk and the infarction zone were determined by a computerized planimetric method using the ImageJ software (Wayne Rasband, Research Services Branch of the National Institute of Mental Health, NIH). The infarct size is expressed as a percentage of the size of the AAR, i.e., as the ratio infarct size/area at risk.

2.4. Pharmacological Agents

Opioid receptor antagonists and inhibitors were administered intravenously 10 min prior to reperfusion. The selective δ2 OR agonist deltorphin II was administered intravenously 5 min prior to reperfusion at a dose of 0.12 mg/kg [5]. The choice of the doses of OR antagonists and the administration schedule were based on our previously published data [4,5]. Naltrexone, which is an antagonist of all types of OR, was used at a dose of 5 mg/kg [5]. The non-selective OR antagonist naloxone methiodide, which does not penetrate the blood–brain barrier, was injected at a dose of 5 mg/kg [5]. The selective peptide δ OR antagonist TIPP [ψ] was administered at a dose of 1 mg/kg [5]. The selective δ2 OR antagonist naltriben mesylate was used at a dose of 0.3 mg/kg [5].
We used inhibitors of kinases and other test substances which are involved in the regulation of cardiac tolerance to I/R. L-NAME, an inhibitor of NO-synthase (NOS), was injected at a dose of 10 mg/kg [8]. Chelerythrine was used at a dose of 5 mg/kg to inhibit PKC [5]. Rottlerin, a PKCδ inhibitor, was injected at a dose of 0.3 mg/kg [9]. The ERK1/2-kinase inhibitor PD-098059 was injected at a dose of 0.5 mg/kg [10]. The Janus 2 kinase inhibitor AG490 was used at a dose of 3 mg/kg [11]. Wortmannin, a PI3-kinase inhibitor, was administered at a dose of 0.025 mg/kg [12]. The AMP-activated protein kinase (AMPK) inhibitor compound C was injected at a dose of 0.25 mg/kg [13]. The NO-sensitive soluble guanylyl cyclase inhibitor ODQ was injected at a dose of 1 mg/kg [14]. The PKA inhibitor H-89 was administered at a dose of 10 µg/kg [15]. Glibenclamide was used to block the KATP channel at a dose of 1 mg/kg [16]. The mitochondrial KATP channel inhibitor 5-hydroxydecanoate (5-HD) was administered at a dose of 5 mg/kg, and the sarcolemmal KATP channel inhibitor HMR 1098 was injected at a dose of 3 mg/kg [17,18]. The BKCa channel inhibitor paxilline was administered at a dose of 1.5 mg/kg [19]. The MPTP opener atractyloside was administered at a dose of 5 mg/kg [15]. The preferential OH scavenger 2-mercaptopropionyl glycine (MPG) was injected at a dose of 20 mg/kg [20]. The superoxide radical scavenger Tempol was used at a dose of 30 mg/kg [21].
All used inhibitors and blockers were administered intravenously 10 min before reperfusion.
The peptides deltorphin II and TIPP[ψ] were provided free by PolyPeptide Laboratories (San Diego, CA). HMR 1098 was synthesized and provided by Sanofi-Aventis Deutschland GmbH (Frankfurt, Germany). Chloralose, 2,3,5-triphenyl tetrazolium chloride, L-NAME, MPG, naltrexone, naloxone methiodide, rottlerin, glibenclamide, 5-HD were purchased from Sigma-Aldrich (St. Louis, MO, USA). Tempol, compound C, ODQ, H-89, paxilline, hydroxypropyl-β-cyclodextrin were purchased from Tocris (Bristol, UK). PD-098059, chelerythrine, AG 490, wortmannin were synthesized and provided free by LCLabs Company (Woburn, MA, USA). The ELISA kits (CEA003Ge cAMP, CEA577Ge cGMP) were purchased from Cloud-Clone Corporation (Wuhan, China). We purchased atractyloside from MedChemExpress (Princeton, NJ, USA). The water-insoluble compounds were dissolved in 0.1 mL of DMSO and then diluted in 0.9 mL of 20% hydroxypropyl-β-cyclodextrin.

2.5. Experimental Protocol

The experiments performed in 528 rats which were divided into 44 groups: 1—control (45 min of ischemia and 2 h reperfusion); 2—naltrexone; 3—naloxone methiodide; 4—TIPP[ψ]; 5—naltriben mesylate; 5—deltorphin II; 6—deltorphin II + naltrexone; 7—deltorphin II + naloxone methiodide; 8—deltorphin II + TIPP[ψ]; 9—deltorphin II + naltriben mesylate; 10—chelerythrine; 11—rottlerin; 12—wortmannin; 13—PD98059; 14—H-89; 15—AG490; 16—L-NAME; 17—compound C; 18—ODQ; 19—glibenclamide; 20—5-HD; 21—HMR1098; 22—atractyloside; 23—paxilline; 24—MPG; 25—tempol; 26—deltorphin II + chelerythrine; 27—deltorphin II + rottlerin; 28—deltorphin II + wortmannin; 29—deltorphin II + PD98059; 30—deltorphin II + H-89; 31—deltorphin II + AG490; 32—deltorphin II + L-NAME; 33—deltorphin II + Compound C; 34—deltorphin II + ODQ; 35—deltorphin II + glibenclamide; 36—deltorphin II + 5-HD; 37—deltorphin II + HMR1098; 38—deltorphin II + atractyloside; 39—deltorphin II + paxilline; 40—deltorphin II + MPG; 41—deltorphin II + tempol; 42—control (cAMP and cGMP level); 43—I/R (cAMP and cGMP level); 44—deltorphin II (cAMP and cGMP level). Each experimental group included 12 rats.

2.6. Detection of Cyclic Nucleotides

The myocardial tissue was frozen in liquid nitrogen. The extraction of cyclic nucleotides was carried out as we previously described [22]. The cAMP and cGMP levels were determined in in the area at risk 15 min after the onset of reperfusion with deltorphin II and without deltorphin II and also in the left ventricle of rats without coronary artery occlusion. The cAMP and cGMP levels were determined in myocardial homogenates using standard commercial ELISA kits (CEA003Ge cAMP, CEA577Ge cGMP) from Cloud-Clone Corporation (Wuhan, China).

2.7. Statistical Analysis

The results are expressed as mean ± standard error of the mean (SEM) from the indicated number of experiments. Data analysis was performed using STATISTICA 13. The homogeneity of the variances was assessed by the Levene’s test. To identify statistically significant differences between independent groups and a single control, we used one-way ANOVA followed by Dunnett’s post hoc test. To identify statistically significant differences in dependent groups, we used one-way repeated-measures ANOVA followed by Dunnett’s post hoc test. To identify statistically significant differences in independent groups, we used one-way ANOVA followed by Bonferroni post hoc test. All the statistical hypotheses were accepted when the significance level was less than p < 0.05.

3. Results

It was demonstrated that deltorphin II and opioid receptor antagonists had no effect on the hemodynamics (Table 1). Most inhibitors also had no effect on the hemodynamics in doses utilized (Table 1). The Janus 2 kinase inhibitor AG490 decreased the heart rate (Table 1). The NO synthase inhibitor L-NAME decreased the heart rate and increased the systolic blood pressure (Table 1).
None of the used OR antagonists had an effect on the infarct size/area at risk ratio (Figure 1).
The selective δ2 opioid receptor agonist deltorphin II decreased the infarct size by 37% (Figure 2).
Pretreatment with the non-selective OR antagonist naltrexone and naloxone methiodide which does not cross the blood–brain barrier (BBB) promoted the removal of the infarct-reducing effect of deltorphin II (Figure 2). The selective δ OR antagonist TIPP (ψ) and the selective δ2 OR antagonist naltriben also contributed to the removal of the infarct-reducing effect of deltorphin II (Figure 2).
Pretreatment with the PKC inhibitor chelerythrine or the selective PKCδ inhibitor rottlerin promoted the elimination of the infarct-sparing effect of deltorphin II (Figure 3).
Pretreatment with the PI3-kinase inhibitor wortmannin promoted the disappearance of the infarct-reducing effect of deltorphin II (Figure 3). The ERK1/2-kinase inhibitor PD98059 contributed to the removal of the infarct-limiting effect of deltorphin II (Figure 3). The PKA inhibitor H-89 did not eliminate the cardioprotective effect of deltorphin II (Figure 3). The Janus 2 kinase inhibitor AG490 also did not eliminate the infarct-limiting effect of deltorphin II (Figure 3). The NOS inhibitor L-NAME and the AMPK inhibitor compound C did not reverse the infarct-reducing effect of deltorphin II (Figure 3). The NO-sensitive soluble guanylyl cyclase inhibitor ODQ promoted the disappearance of the cardioprotective effect of deltorphin II (Figure 3).
The non-selective KATP channel blocker glibenclamide contributed to the removal of the infarct-sparing effect of deltorphin II (Figure 4).
The mitochondrial KATP channel blocker 5-HD did not reverse the infarct-reducing effect of deltorphin II (Figure 4). In contrast, the sarcolemmal KATP channel blocker HMR 1098 contributed to the removal of the infarct-limiting effect of deltorphin II (Figure 4). The MPTP opener atractyloside also promoted the elimination of the cardioprotective effect of deltorphin II (Figure 4). The BKCa channel inhibitor paxilline did not abolish the infarct-reducing effect of deltorphin II (Figure 4).
We studied the role of ROS in deltorphin II-induced cardioprotection. It was determined that the preferential OH scavenger MPG did not reverse the infarct-reducing effect of deltorphin II (Figure 4). The superoxide scavenger Tempol did not abolish the cardioprotective effect of deltorphin II (Figure 4).
It should be noted that all inhibitors had no effect on the infarct size/area at risk ratio (Figure 5 and Figure 6).
Reperfusion caused a decrease in the cAMP level and an increase in the cGMP level in the area at risk (Table 2). Deltorphin II promoted a five-fold increase in the cGMP level in the area at risk compared to the control group of animals with I/R of the heart.

4. Discussion

It was shown that the infarct-reducing effect of deltorphin II is mediated via the activation of peripheral δ2 OR. These data are consistent with our previously published data [4]. This δ2 OR appears to be localized on the sarcolemma of cardiomyocytes, because deltorphin II increases the resistance of isolated cardiomyocytes to anoxia/reoxygenation via the stimulation of δ2 OR [4].
It was found that PKCδ is involved in the infarct-sparing effect of deltorphin II. This result is consistent with our previously published data indicating that the cardioprotective effect of opioids was associated with protein kinase activation in I/R of the heart [4]. Consequently, PKCδ is involved in the cardioprotective effect of opioids during reperfusion.
It was also demonstrated that the infarct-reducing effect of deltorphin II is associated with the activation of PI3-kinase and ERK1/2-kinase. The obtained result is consistent with previously published data of the Gross’ group showing that PI3-kinase and ERK1/2-kinase participate in the cardioprotective effect of the κ OR agonist U-50488 at reperfusion [23].
There is evidence that PKA, AMPK, and Janus 2 are involved in morphine-induced preconditioning [11,15,24]. However, we demonstrated that AMP-activated protein kinase, Janus2, and PKA did not participate in the infarct-reducing effect of deltorphin II. Consequently, it could be proposed that the molecular mechanism of the cardioprotective effect of morphine differs from that of the cardioprotective effect of deltorphin II. Morphine, unlike deltorphin II, readily crosses the blood–brain barrier, thereby the cardioprotective effect of morphine could be mediated via stimulation of central opioid receptors. Therefore, the molecular mechanism of the cardioprotective effect of morphine could be different from the mechanism of deltorphin-triggered cardioprotection. Morphine was used before ischemia [11,15,24], thereby we cannot rule out the possibility that the molecular mechanism of opioid-induced preconditioning could be different from the mechanism of opioid-induced postconditioning.
It was previously reported that NOS is involved in the cardioprotective effect of the opioid Eribis peptide 94 [25]. However, our data indicate that NOS is not involved in the cardioprotective effect of deltorphin II.
It is generally accepted that the mitochondrial KATP channel is involved in the infarct-limiting effect of opioids when used before cardiac I/R [5,26] and at reperfusion [23]. However, the results of our study indicate that the sarcolemmal KATP channel can also participate in the infarct-limiting effect of deltorphin II in reperfusion. It was previously reported that MPTP and the BKCa channel are involved in the cardioprotective effect of the κ OR agonist U-50488 [27]. We also obtained evidence that the infarct-reducing effect of deltorphin II is mediated via MPTP closing. The BKCa channel did not participate in the cardioprotective effect of deltorphin II.
Reactive oxygen species could be intracellular signaling molecules [28]. There is evidence that ROS are involved in the cardioprotective effect of the δ OR agonist SNC-121 in reperfusion [20]. However, our studies demonstrated that the infarct-sparing effect of deltorphin II in reperfusion does not involve ROS. Consequently, the signaling mechanism of the cardioprotective effect of deltorphin II could be different from the signaling mechanism of the protective effect of SNC-121. It is possible that the cardioprotective effect of SNC-121 is the result of central OR stimulation and the cardioprotective effect of deltorphin II is the result of peripheral δ2 OR activation; therefore, the molecular mechanisms of their effects could be different.
We previously reported that the non-selective peptide δ OR agonist dalargin can increase the cGMP level in the area at risk during ischemia [22]. Deltorphin II had a similar effect at reperfusion. It has been reported that there are two main source of cGMP formation in cardiomyocyte: natriuretic peptide receptor and soluble NO-sensitive guanylyl cyclase [29]. Pretreatment with the soluble NO-sensitive guanylyl cyclase inhibitor ODQ completely removed the cardioprotective effect of deltorphin II. This was an unexpected result, since NOS inhibition did not affect the infarct-sparing effect of deltorphin II. It should be noted that soluble guanylyl cyclase can be activated not only by NO but also by CO. It is possible that deltorphin II activated heme oxygenase-1 and increased CO production and CO stimulation of guanylyl cyclase, leading to cardioprotection. There is also indirect evidence that ODQ can inhibit natriuretic peptide-sensitive guanylyl cyclase [30].
It was found that deltorphin II contributed to a reduction in the myocardial cAMP level in reperfusion (Table 2). This result is in agreement with previous findings that the peptide δ OR agonist dalargin reduced the myocardial cAMP level in ischemia [22]. There are data that opioid receptors are coupled with Gi/o proteins [31]. The stimulation of Gi/o proteins promotes the inhibition of the adenylyl cycle [31]. It could be proposed that deltorphin II promoted the inhibition of the adenylyl cycle via the activation of Gi/o proteins.

5. Conclusions

Our study showed that the cardioprotective effect of deltorphin II in reperfusion is mediated via the activation of peripheral δ2 OR, which is probably localized in cardiomyocytes. The infarct-reducing effect of deltorphin II is mediated via the activation of PKCδ, PI3-kinase, ERK1/2-kinase, sarcolemmal KATP channel opening, MPTP closing. PKA, Janus 2, AMP-activated protein kinase, NOS, ROS, the BKCa channel, and the mitochondrial KATP channel are not involved in the infarct-limiting effect of deltorphin II in reperfusion. We also demonstrated that guanylyl cyclase plays an important role in the cardioprotective effect of deltorphin II in reperfusion.

Author Contributions

Conceptualization, S.V.P.; methodology, L.N.M.; formal analysis, V.N.A.; investigation and visualization, A.V.M. and B.K.K.; data curation, L.N.M. and N.V.N.; writing—original draft preparation, N.R.P. and N.S.; writing—review and editing, L.N.M. and F.F.; supervision, S.V.P. and N.V.N.; project administration, S.V.P. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Russian Science Foundation Grant 22-15-00048. The section dedicated to protein kinase C is within the framework of state assignments 122020300042-4.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors are grateful to Kevin Gormley (NIDA, NIH, Bethesda) for providing deltorphin II and TIPP (ψ).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Olier, I.; Sirker, A.; Hildick-Smith, D.; Kinnaird, T.; Ludman, P.; de Belder, M.A.; Baumbach, A.; Byrne, J.; Rashid, M.; Curzen, N.; et al. British cardiovascular intervention society and the national institute for cardiovascular outcomes research. Association of different antiplatelet therapies with mortality after primary percutaneous coronary intervention. Heart 2018, 104, 1683–1690. [Google Scholar] [CrossRef] [PubMed]
  2. McCartney, P.J.; Berry, C. Redefining successful primary PCI. Eur. Heart J. Cardiovasc. Imaging 2019, 20, 133–135. [Google Scholar] [CrossRef] [PubMed]
  3. Ong, S.B.; Samangouei, P.; Kalkhoran, S.B.; Hausenloy, D.J. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J. Mol. Cell Cardiol. 2015, 78, 23–34. [Google Scholar] [CrossRef] [PubMed]
  4. Maslov, L.N.; Mukhomedzyanov, A.V.; Tsibulnikov, S.Y.; Suleiman, M.S.; Khaliulin, I.; Oeltgen, P.R. Activation of peripheral δ2-opioid receptor prevents reperfusion heart injury. Eur. J. Pharmacol. 2021, 907, 174302. [Google Scholar] [CrossRef] [PubMed]
  5. Maslov, L.N.; Lishmanov, Y.B.; Oeltgen, P.R.; Barzakh, E.I.; Krylatov, A.V.; Govindaswami, M.; Brown, S.A. Activation of peripheral δ2 opioid receptors increases cardiac tolerance to ischemia/reperfusion injury: Involvement of protein kinase C, NO-synthase, KATP channels and the autonomic nervous system. Life Sci. 2009, 84, 657–663. [Google Scholar] [CrossRef] [PubMed]
  6. Maslov, L.N.; Naryzhnaia, N.V.; Tsibulnikov, S.Y.; Kolar, F.; Zhang, Y.; Wang, H.; Gusakova, A.M.; Lishmanov, Y.B. Role of endogenous opioid peptides in the infarct size-limiting effect of adaptation to chronic continuous hypoxia. Life Sci. 2013, 93, 373–379. [Google Scholar] [CrossRef]
  7. Neckár, J.; Sźárszoi, O.; Herget, J.; Ostádal, B.; Kolár, F. Cardioprotective effect of chronic hypoxia is blunted by concomitant hypercapnia. Physiol. Res. 2003, 52, 171–175. [Google Scholar]
  8. Ebrahim, Z.; Yellon, D.M.; Baxter, G.F. Bradykinin elicits “second window” myocardial protection in rat heart through an NO-dependent mechanism. Am. J. Physiol. Heart Circ. Physiol. 2001, 281, 1458–1464. [Google Scholar] [CrossRef]
  9. Zatta, A.J.; Kin, H.; Lee, G.; Wang, N.; Jiang, R.; Lust, R.; Reeves, J.G.; Mykytenko, J.; Guyton, R.A.; Zhao, Z.Q.; et al. Infarct-sparing effect of myocardial postconditioning is dependent on protein kinase C signaling. Cardiovasc. Res. 2006, 70, 315–334. [Google Scholar] [CrossRef]
  10. Lasley, R.D.; Keith, B.J.; Kristo, G.; Yoshimura, Y.; Mentzer, R.M., Jr. Delayed adenosine A1 receptor preconditioning in rat myocardium is MAPK dependent but iNOS independent. Am. J. Physiol. Heart Circ. Physiol. 2005, 289, 785–791. [Google Scholar] [CrossRef] [Green Version]
  11. Gross, E.R.; Hsu, A.K.; Gross, G.J. The JAK/STAT pathway is essential for opioid-induced cardioprotection: JAK2 as a mediator of STAT3, Akt, and GSK-3β. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, 827–834. [Google Scholar] [CrossRef] [PubMed]
  12. Fettiplace, M.R.; Kowal, K.; Ripper, R.; Young, A.; Lis, K.; Rubinstein, I.; Bonini, M.; Minshall, R.; Weinberg, G. Insulin signaling in bupivacaine-induced cardiac toxicity: Sensitization during recovery and potentiation by lipid emulsion. Anesthesiology 2016, 124, 428–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hong, L.; Sun, Y.; An, J.Z.; Wang, C.; Qiao, S.G. Sevoflurane preconditioning confers delayed cardioprotection by upregulating AMP-activated protein kinase levels to restore autophagic flux in ischemia-reperfusion rat hearts. Med. Sci. Monit. 2020, 26, e922176. [Google Scholar] [CrossRef] [PubMed]
  14. Schindler, U.; Strobel, H.; Schönafinger, K.; Linz, W.; Löhn, M.; Martorana, P.A.; Rütten, H.; Schindler, P.W.; Busch, A.E.; Sohn, M.; et al. Biochemistry and pharmacology of novel anthranilic acid derivatives activating heme-oxidized soluble guanylyl cyclase. Mol. Pharmacol. 2006, 69, 1260–1268. [Google Scholar] [CrossRef] [Green Version]
  15. Dorsch, M.; Behmenburg, F.; Raible, M.; Blase, D.; Grievink, H.; Hollmann, M.W.; Heinen, A.; Huhn, R. Morphine-induced preconditioning: Involvement of protein kinase A and mitochondrial permeability transition pore. PLoS ONE 2016, 11, e0151025. [Google Scholar] [CrossRef]
  16. Nithipatikom, K.; Moore, J.M.; Isbell, M.A.; Falck, J.R.; Gross, G.J. Epoxyeicosatrienoic acids in cardioprotection: Ischemic versus reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, 537–542. [Google Scholar] [CrossRef] [Green Version]
  17. Fryer, R.M.; Eells, J.T.; Hsu, A.K.; Henry, M.M.; Gross, G.J. Ischemic preconditioning in rats: Role of mitochondrial KATP channel in preservation of mitochondrial function. Am. J. Physiol. Heart Circ. Physiol. 2000, 278, 305–312. [Google Scholar] [CrossRef]
  18. Fryer, R.M.; Hsu, A.K.; Nagase, H.; Gross, G.J. Opioid-induced cardioprotection against myocardial infarction and arrhythmias: Mitochondrial versus sarcolemmal ATP-sensitive potassium channels. J. Pharmacol. Exp. Ther. 2000, 294, 451–457. [Google Scholar]
  19. Smith, B.L.; McLeay, L.M.; Embling, P.P. Effect of the mycotoxins penitrem, paxilline and lolitrem B on the electromyographic activity of skeletal and gastrointestinal smooth muscle of sheep. Res. Vet. Sci. 1996, 62, 111–116. [Google Scholar] [CrossRef]
  20. Tsutsumi, Y.M.; Yokoyama, T.; Horikawa, Y.; Roth, D.M.; Patel, H.H. Reactive oxygen species trigger ischemic and pharmacological postconditioning: In vivo and in vitro characterization. Life Sci. 2007, 81, 1223–1227. [Google Scholar] [CrossRef] [Green Version]
  21. Pınar, N.; Kaplan, M.; Özgür, T.; Özcan, O. Ameliorating effects of tempol on methotrexate-induced liver injury in rats. Biomed. Pharmacother. 2018, 102, 758–764. [Google Scholar] [CrossRef] [PubMed]
  22. Maslov, L.N.; Lishmanov, Y.B. The anti-arrhythmic effect of D-Ala2, Leu5,Arg6-enkephalin and its possible mechanism. Int. J. Cardiol. 1993, 40, 89–94. [Google Scholar] [CrossRef] [PubMed]
  23. Peart, J.N.; Gross, E.R.; Reichelt, M.E.; Hsu, A.; Headrick, J.P.; Gross, G.J. Activation of kappa-opioid receptors at reperfusion affords cardioprotection in both rat and mouse hearts. Basic. Res. Cardiol. 2008, 103, 454–463. [Google Scholar] [CrossRef] [PubMed]
  24. Li, L.; Zhang, H.; Li, T.; Zhang, B. Involvement of adenosine monophosphate-activated protein kinase in morphine-induced cardioprotection. J. Surg. Res. 2011, 169, 179–187. [Google Scholar] [CrossRef]
  25. Gross, G.J.; Hsu, A.; Nithipatikom, K.; Pfeiffer, A.W.; Bobrova, I.; Bissessar, E. Acute and chronic cardioprotection by the enkephalin analogue, Eribis peptide 94, is mediated via activation of nitric oxide synthase and adenosine triphosphate-regulated potassium channels. Pharmacology 2012, 90, 110–116. [Google Scholar] [CrossRef] [Green Version]
  26. Zhang, Y.; Irwin, M.G.; Wong, T.M.; Chen, M.; Cao, C.M. Remifentanil preconditioning confers cardioprotection via cardiac kappa- and delta-opioid receptors. Anesthesiology 2005, 102, 371–378. [Google Scholar] [CrossRef]
  27. Cao, C.M.; Chen, M.; Wong, T.M. The KCa channel as a trigger for the cardioprotection induced by kappa-opioid receptor stimulation—Its relationship with protein kinase C. Br. J. Pharmacol. 2009, 145, 984–991. [Google Scholar] [CrossRef] [Green Version]
  28. Krylatov, A.V.; Maslov, L.N.; Voronkov, N.S.; Boshchenko, A.A.; Popov, S.V.; Gomez, L.; Wang, H.; Jaggi, A.S.; Downey, J.M. Reactive oxygen species as intracellular signaling molecules in the cardiovascular system. Curr. Cardiol. Rev. 2018, 14, 290–300. [Google Scholar] [CrossRef]
  29. Krylatov, A.V.; Tsibulnikov, S.Y.; Mukhomedzyanov, A.V.; Boshchenko, A.A.; Goldberg, V.E.; Jaggi, A.S.; Erben, R.G.; Maslov, L.N. The role of natriuretic peptides in the regulation of cardiac tolerance to ischemia/reperfusion and postinfarction heart remodeling. J. Cardiovasc. Pharmacol. Ther. 2021, 26, 131–148. [Google Scholar] [CrossRef]
  30. Yang, X.M.; Philipp, S.; Downey, J.M.; Cohen, M.V. Atrial natriuretic peptide administered just prior to reperfusion limits infarction in rabbit hearts. Basic Res. Cardiol. 2006, 10, 311–318. [Google Scholar] [CrossRef]
  31. Maslov, L.N.; Khaliulin, I.; Oeltgen, P.R.; Naryzhnaya, N.V.; Pei, J.M.; Brown, S.A.; Lishmanov, Y.B.; Downey, J.M. Prospects for creation of cardioprotective and antiarrhythmic drugs based on opioid receptor agonists. Med. Res. Rev. 2016, 36, 871–923. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of opioid receptor antagonists on the infarct size/area at risk ratio (Y-axis). Data represent the means ± SEM. Each study group included 12 rats.
Figure 1. Effect of opioid receptor antagonists on the infarct size/area at risk ratio (Y-axis). Data represent the means ± SEM. Each study group included 12 rats.
Membranes 13 00063 g001
Figure 2. Involvement of opioid receptors in the mechanism of the infarct-limiting effect of deltorphin II (Y-axis). Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Figure 2. Involvement of opioid receptors in the mechanism of the infarct-limiting effect of deltorphin II (Y-axis). Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Membranes 13 00063 g002
Figure 3. Involvement of kinases and NOS in the infarct-limiting effect of deltorphin II (Y-axis). 1—Control; 2—Deltorphin II; 3—Deltorphin II + Chelerythrine; 4—Deltorphin II + Rottlerin; 5—Deltorphin II + Wortmannin; 6—Deltorphin II + PD98059; 7—Deltorphin II + H-89; 8—Deltorphin II + AG490; 9—Deltorphin II + L-NAME; 10—Deltorphin II + Compound C; 11—Deltorphin II + ODQ. Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Figure 3. Involvement of kinases and NOS in the infarct-limiting effect of deltorphin II (Y-axis). 1—Control; 2—Deltorphin II; 3—Deltorphin II + Chelerythrine; 4—Deltorphin II + Rottlerin; 5—Deltorphin II + Wortmannin; 6—Deltorphin II + PD98059; 7—Deltorphin II + H-89; 8—Deltorphin II + AG490; 9—Deltorphin II + L-NAME; 10—Deltorphin II + Compound C; 11—Deltorphin II + ODQ. Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Membranes 13 00063 g003
Figure 4. The involvement of KATP channels, MPT pore, the BKCa channel, and ROS in the infarct-reducing effect of deltorphin II (Y-axis). 1—Control; 2—Deltorphin II; 3—Deltorphin II + Glibenclamide; 4—Deltorphin II + 5-HD; 5—Deltorphin II + HMR1098; 6—Deltorphin II + Atractyloside; 7—Deltorphin II + Paxilline; 8—Deltorphin II + MPG; 9—Deltorphin II + Tempol. Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Figure 4. The involvement of KATP channels, MPT pore, the BKCa channel, and ROS in the infarct-reducing effect of deltorphin II (Y-axis). 1—Control; 2—Deltorphin II; 3—Deltorphin II + Glibenclamide; 4—Deltorphin II + 5-HD; 5—Deltorphin II + HMR1098; 6—Deltorphin II + Atractyloside; 7—Deltorphin II + Paxilline; 8—Deltorphin II + MPG; 9—Deltorphin II + Tempol. Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the deltorphin II group. Each study group included 12 rats.
Membranes 13 00063 g004
Figure 5. Effect of inhibitors of kinases and molecular structures on the infarct size/area at risk ratio (Y-axis). 1—Control; 2—Chelerythrine; 3—Rottlerin; 4—Wortmannin; 5—PD98059; 6—H-89; 7—AG490; 8—L-NAME; 9—Compound C; 10—ODQ. Data represent the means ± SEM. Each study group included 12 rats.
Figure 5. Effect of inhibitors of kinases and molecular structures on the infarct size/area at risk ratio (Y-axis). 1—Control; 2—Chelerythrine; 3—Rottlerin; 4—Wortmannin; 5—PD98059; 6—H-89; 7—AG490; 8—L-NAME; 9—Compound C; 10—ODQ. Data represent the means ± SEM. Each study group included 12 rats.
Membranes 13 00063 g005
Figure 6. Effect inhibitors of kinases and molecular structures on the infarct size/area at risk ratio (Y-axis). 1—Control; 2—Glibenclamide; 3—5-HD; 4—HMR1098; 5—Atractyloside; 6—Paxilline; 7—MPG; 8—Tempol. Data represent the means ± SEM. Each study group included 12 rats.
Figure 6. Effect inhibitors of kinases and molecular structures on the infarct size/area at risk ratio (Y-axis). 1—Control; 2—Glibenclamide; 3—5-HD; 4—HMR1098; 5—Atractyloside; 6—Paxilline; 7—MPG; 8—Tempol. Data represent the means ± SEM. Each study group included 12 rats.
Membranes 13 00063 g006
Table 1. Hemodynamics data.
Table 1. Hemodynamics data.
Before IschemiaBefore ReperfusionAfter 30 min ReperfusionAfter 2 h Reperfusion
Heart rate (beats/min)
Control365 ± 4359 ± 4352 ± 5342 ± 9
Deltorphin II367 ± 5362 ± 5354 ± 4348 ± 8
Naltrexone359 ± 8353 ± 6348 ± 5340 ± 7
Naloxone methiodide355 ± 6349 ± 4345 ± 5336 ± 6
TIPP [ψ]366 ± 3358 ± 4353 ± 4344 ± 7
Naltriben358 ± 5352 ± 4346 ± 3337 ± 7
L-NAME366 ± 5335 ± 5 *318 ± 4 *304 ± 9 *
Chelerythrine361 ± 4354 ± 3347 ± 5339 ± 6
Rottlerin370 ± 3363 ± 5356 ± 4345 ± 7
PD098059362 ± 4355 ± 4350 ± 5341 ± 8
AG490367 ± 3340 ± 4 *337 ± 5 *321 ± 7 *
Wortmannin360 ± 5354 ± 3349 ± 4339 ± 9
ODQ365 ± 4359 ± 4354 ± 5345 ± 7
H-89367 ± 5361 ± 5356 ± 4344 ± 8
Compound C364 ± 4360 ± 6354 ± 5346 ± 9
Glibenclamide359 ± 4354 ± 3349 ± 5338 ± 6
5-HD368 ± 3363 ± 3358 ± 4347 ± 5
HMR1098362 ± 4356 ± 5350 ± 4336 ± 7
Paxilline357 ± 5352 ± 4345 ± 5335 ± 8
Atractyloside366 ± 4360 ± 4356 ± 3345 ± 5
MPG361 ± 3355 ± 4347 ± 4337 ± 7
Tempol356 ± 5351 ± 4344 ± 3334 ± 8
Mean systolic blood pressure (mmHg)
Control126 ± 5122 ± 3119 ± 4114 ± 6
Deltorphin II122 ± 4119 ± 4115 ± 5109 ± 7
Naltrexone125 ± 3120 ± 5117 ± 3111 ± 5
Naloxone methiodide128 ± 4124 ± 4120 ± 5113 ± 7
TIPP[ψ]124 ± 3119 ± 5116 ± 3111 ± 8
Naltriben120 ± 3117 ± 4113 ± 3106 ± 5
L-NAME123 ± 4145 ± 3 *155 ± 5 *161 ± 6 *
Chelerythrine126 ± 3123 ± 4118 ± 4113 ± 5
Rottlerin125 ± 4121 ± 4116 ± 5111 ± 6
PD098059128 ± 3124 ± 5120 ± 3112 ± 7
AG490127 ± 4140 ± 3 *138 ± 4 *134 ± 5 *
Wortmannin126 ± 5121 ± 4117 ± 3113 ± 7
ODQ122 ± 3117 ± 3114 ± 5108 ± 5
H-89127 ± 4123 ± 4118 ± 5113 ± 8
Compound C124 ± 6121 ± 4119 ± 3114 ± 5
Glibenclamide128 ± 5125 ± 5121 ± 7117 ± 6
5-HD 121 ± 3118 ± 4114 ± 6109 ± 5
HMR1098125 ± 3120 ± 5117 ± 3112 ± 7
Paxilline119 ± 6115 ± 4111 ± 5105 ± 8
Atractyloside128 ± 3123 ± 4118 ± 4113 ± 6
MPG124 ± 4121 ± 3117 ± 4110 ± 7
Tempol121 ± 5116 ± 4112 ± 3107 ± 8
Data represent the means ± SEM. * p < 0.05 vs. the control group.
Table 2. Effect of deltorphin II on the cAMP and cGMP levels in ischemia/reperfusion of the heart.
Table 2. Effect of deltorphin II on the cAMP and cGMP levels in ischemia/reperfusion of the heart.
GroupcAMP (nmol/g)cGMP (nmol/g)
Control18.2 ± 1.19 ± 1.1
I/R13.1 ± 3.9 *24 ± 4.4 *
Deltorphin II11.8 ± 2.8 #44 ± 4.7 *,#
Data represent the means ± SEM. * p < 0.05 vs. the control group; # p < 0.05 vs. the I/R group; I/R, ischemia/reperfusion.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Popov, S.V.; Mukhomedzyanov, A.V.; Maslov, L.N.; Naryzhnaya, N.V.; Kurbatov, B.K.; Prasad, N.R.; Singh, N.; Fu, F.; Azev, V.N. The Infarct-Reducing Effect of the δ2 Opioid Receptor Agonist Deltorphin II: The Molecular Mechanism. Membranes 2023, 13, 63. https://doi.org/10.3390/membranes13010063

AMA Style

Popov SV, Mukhomedzyanov AV, Maslov LN, Naryzhnaya NV, Kurbatov BK, Prasad NR, Singh N, Fu F, Azev VN. The Infarct-Reducing Effect of the δ2 Opioid Receptor Agonist Deltorphin II: The Molecular Mechanism. Membranes. 2023; 13(1):63. https://doi.org/10.3390/membranes13010063

Chicago/Turabian Style

Popov, Sergey V., Alexandr V. Mukhomedzyanov, Leonid N. Maslov, Natalia V. Naryzhnaya, Boris K. Kurbatov, N. Rajendra Prasad, Nirmal Singh, Feng Fu, and Viacheslav N. Azev. 2023. "The Infarct-Reducing Effect of the δ2 Opioid Receptor Agonist Deltorphin II: The Molecular Mechanism" Membranes 13, no. 1: 63. https://doi.org/10.3390/membranes13010063

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop